Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Journal of Clinical Virology 21 (2001) 197– 212

www.elsevier.com/locate/jcv

Resistance of human immunodeficiency virus type 1 to


reverse transcriptase and protease inhibitors: genotypic and
phenotypic testing
J. Gerardo Garcı́a-Lerma, Walid Heneine *
HIV and Retro6irology Branch, Di6ision of AIDS, STD, and TB Laboratory Research, National Center for Infectious Diseases,
Centers for Disease Control and Pre6ention, Atlanta, GA 30333, USA

Abstract

Treatment of HIV-1-infected persons with antiretroviral drugs including reverse transcriptase (RT) and protease
inhibitors has significantly reduced the rate of HIV and AIDS-related morbidity and mortality. However, these
treatments can select for drug-resistant viruses which are associated with poor virologic responses to the antiretroviral
therapies and loss of clinical benefit. Drug resistance is conferred by single or several amino acid changes in the pol
gene. These mutations can be classified as primary when they directly confer reduced drug susceptibility, or secondary
when their influence is primarily on replication capabilities of resistant viruses. Both genotypic and phenotypic
methods are used for drug resistance testing. Genotypic assays detect resistance-related mutations by sequence
analysis or point mutations assays. Phenotypic testing measures drug susceptibility of patient-derived viruses in
culture assays. Viruses can be conventionally isolated from peripheral blood lymphocytes, or generated more rapidly
through recombination of plasma-derived RT/protease sequences and modified HIV-1 vectors. Phenotypic testing
provides direct evidence of resistance, is easy to interpret, but is laborious and expensive. In contrast, genotypic
testing provides indirect evidence of resistance, is relatively faster and cheaper, but some complex mutation patterns
may be difficult to interpret. Non-culture based phenotypic assays that measure susceptibility of RT activity in plasma
to RT inhibitors have been described recently, and provide new tools for rapid phenotypic testing. Resistance testing
is currently recommended to help guide the choice of new regimens after treatment failure and for guiding therapy
in pregnant women. © 2001 Elsevier Science B.V. All rights reserved.

Keywords: HIV-1 drug resistance; Phenotype; Genotype

1. Introduction munodeficiency virus type 1 (HIV-1) (Mitsuya et


al., 1985), drug therapy has been widely used in
Since the discovery of zidovudine (ZDV) as an the treatment of AIDS. Combination of antiretro-
effective antiretroviral agent against human im- viral drugs targeting the reverse transcriptase
(RT) and/or protease of HIV-1 can suppress
* Corresponding author. Tel.: + 1-404-6390218; fax: + 1- plasma levels of the virus and delay disease pro-
404-6391174. gression in HIV-1-infected persons, thus reducing

1386-6532/01/$ - see front matter © 2001 Elsevier Science B.V. All rights reserved.
PII: S 1 3 8 6 - 6 5 3 2 ( 0 0 ) 0 0 1 6 3 - 3
198 J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212

the rate of HIV and AIDS-related morbidity and HIV-1 RT include nucleoside and non-nucleoside
mortality (Gulick et al., 1997; Hammer et al., RT inhibitors. The nucleoside RT inhibitors (NR-
1997). Despite their ability to suppress virus repli- TIs) class of compounds share a similar mecha-
cation for extended periods of time, current an- nism of action and require intracellular activation
tiretroviral therapies have thus far been unable to by phosphorylation to the 5%-triphosphate form
eradicate HIV-1 from infected persons (Pierson et (Arts and Wainberg, 1996). The triphosphate
al., 2000). form of NRTIs inhibits HIV-1 RT through com-
The clinical benefit of antiretroviral therapy is petitive inhibition with the triphosphate form of
compromised by the emergence of drug-resistant the native deoxynucleoside, and chain termination
variants of HIV-1. Emergence of drug-resistant of elongating DNA (Arts and Wainberg, 1996).
HIV-1 is due to the ability of the virus to escape The six NRTIs approved for treatment are ZDV,
under drug-selective pressures. Virus escape is a didanosine (ddI), zalcitabine (ddC), stavudine
consequence of the population structure of HIV-1 (d4T), lamivudine (3TC) and abacavir (1592U89)
which consists of a complex distribution of related (Mitsuya et al., 1985; Mitsuya and Broder, 1986;
but not identical HIV-1 genomes known as qua- Lin et al., 1987; Yarchoan et al., 1989; Coates et
sispecies (Domingo et al., 1995, 1997, 1998). The al., 1992; Daluge et al., 1997).
quasispecies distribution results from the absence The non-nucleoside RT inhibitors (NNRTIs)
of proofreading-repair activities during RNA-de- are structurally and chemically dissimilar com-
pendent DNA synthesis by the viral RT. Calcula- pounds that are potent inhibitors of HIV-1 RT.
tions of the mutation rate for HIV-1 predict that In contrast to NRTIs, the NNRTIs do not require
each newly copied genome differs from the metabolic activation to manifest their antiviral
parental virus on average by a single nucleotide
(Mansky and Temin, 1995). If an estimated 1010 Table 1
virions are produced daily in an HIV-1-infected Antiretroviral drugs currently approved for treatment of HIV-
person (Perelson et al., 1996) and each genome 1-infected persons
contains an average of 1 mutation, every possible Generic name Trade name FDA approval
single mutation associated with drug resistance
may be generated daily. Drug resistance muta- Nucleoside RT inhibitors
tions have been identified in the RT gene from Zidovudine (ZDV) Retrovir® 1986
patients who have not received antiretroviral Didanosine (ddl) Videx® 1991
Zalcitabine (ddC) Hivid® 1992
drugs (Nájera et al., 1994, 1995; de Jong et al., Stavudine (d4T) Zerit® 1994
1996). Estimates based on the rate of emergence Lamivudine (3TC) Epivir® 1995
of nevirapine resistance in previously untreated Abacavir (1592U89) Ziagen® 1998
persons indicate that approximately 1 in 1000 Non-nucleoside RT inhibitors
genomes carry the Y181C mutation associated Nevirapine Viramune® 1996
with nevirapine resistance before treatment with (BI-RG-587)
nevirapine (Havlir et al., 1996). Therefore, pre-ex- Delavirdine Rescriptor® 1997
(U-90152)
isting viruses with resistance mutations may be-
Efavirenz Sustiva™, 1998
come rapidly selected by antiretroviral drugs. (DMP 266) Stocrin™
Protease inhibitors
Indinavir (MK 639) Crixivan® 1996
2. Inhibitors of HIV-1 reverse transcriptase and Ritonavir (ABT 538) Norvir® 1996
protease Saquinavir Invirase®, 1995, 1997
(RO 31-8959) Fortovase®
To date, a total of 14 antiretroviral drugs have Nelfinavir Viracept® 1997
(AG 1343)
been approved for treatment of HIV-1-infected Amprenavir Agenerase® 1999
persons (Table 1). These drugs target either the (141W94)
RT or the protease of the virus. Inhibitors of
J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212 199

effects (Spence et al., 1995; Joly and Yeni, 1999). Table 2


Mutations associated with resistance to HIV-1 RT inhibitors
Inhibition of HIV-1 RT by NNRTIs occurs by
non competitive binding to conserved residues of RT inhibitor Primary mutations Secondary
the p66 subunit of the RT, which alters the con- mutations
formational structure and modify the active site of
the enzyme (Spence et al., 1995; Maga et al., 1997; Nucleoside RT inhibitors
Zidovudine K70R, T215Y/F M41L, D67N,
Joly and Yeni 1999). These compounds are spe-
L210W, K219Q
cific for HIV-1 group M isolates and do not have Didanosine L74V K65R, M184V/I
a significant antiviral effect on HIV-1 group O, Zalcitabine K65R, T69D,
HIV-2, or SIV isolates (Descamps et al., 1995, L74V, M184V/I
1997; Witvrouw et al., 1999). The three NNRTIs Lamivudine E44D, V 118I,
M184V
approved are nevirapine, delavirdine, and efa-
Stavudine V75T
virenz (Wu et al., 1991; Dueweke et al., 1993b; Abacavir K65R, L74V, M41L, D67N,
Young et al., 1995). M184V K70R, Y115F,
The HIV-1 protease inhibitors class of com- L210W, K219Q
pounds are peptidomimetic that bind to the active Multinucleoside Q151M A62V, V75I, F77L,
resistance (a) F116Y
site of the viral protease enzyme. Unlike RT
Multinucleoside T69S-SA/G/S M41L, A62V,
inhibitors, protease inhibitors block the produc- resistance (b) D67N, K70R,
tion of infectious virus from infected cells by L210W, T215Y,
inhibiting the cleavage of precursor polyproteins K219Q
necessary to produce infectious virus particles. Non-nucleoside RT inhibitors
Therefore, virus particles produced by infected Nevirapine K103N, V106A, L100I
cells treated with protease inhibitors contain un- V108I, Y181C,
processed precursors and are non-infectious (Kohl Y188C, G190A
Delavirdine K103N, Y181C P236L
et al., 1988; Peng et al., 1989). The five currently
Efavirenz K103N, Y188L, L100I, V108I,
approved protease inhibitors are saquinavir, indi- G190A P225H
navir, ritonavir, nelfinavir, and amprenavir
(Roberts et al., 1990; Ho et al., 1994; Vacca et al.,
1994; Parteledis et al., 1995; Kaldor et al., 1997). A62V, V75I, F77L, and F116Y mutations in-
crease replication capabilities and level of resis-
tance in viruses carrying the Q151M mutation
3. Mutations associated with drug resistance (Maeda et al., 1998; Kosalaraksa et al., 1999), and
the L63P mutation compensates for the impair-
Mutations associated with resistance can be ment of fitness caused by the saquinavir-selected
classified as primary or secondary. Primary muta- L90M mutation (Martı́nez-Picado et al., 1999). A
tions usually appear early, are relatively specific list of primary and secondary mutations associ-
for each drug, and decrease drug susceptibility. ated with resistance to RT and protease inhibitors
Secondary mutations, also called compensatory is shown in Table 2 and Table 3, respectively.
mutations, usually accumulate in viral genomes
that already have some primary mutations. These
secondary mutations confer little or no reduction 3.1. Resistance to NRTIs
in drug susceptibility by themselves, restore repli-
cation capabilities of virus carrying primary muta- Resistance to NRTIs may result from single
tions, and may also increase the level of resistance mutations or accumulation of several mutations
in such viruses. For instance, the L210W muta- in the pol gene. For instance, the M184V muta-
tion increases the level of ZDV resistance of tion that is associated with resistance to 3TC can
viruses that harbor other ZDV resistant mutations alone reduce the susceptibility for this drug \
(Harrigan et al., 1996; Hooker et al., 1996), the 100-fold (Boucher et al., 1993; Schinazi et al.,
200 J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212

1993; Tisdale et al., 1993). Other mutations such Resistance mutations to a particular NRTI can
as E44D or V118I can also confer moderate (4– affect the susceptibility of the virus to other
50-fold) levels of 3TC resistance in a background NRTI. For instance, the M184V or L74V muta-
of ZDV resistance mutations (Hertogs et al., tions selected during 3TC and ddI treatment,
2000). High level resistance to ZDV requires the respectively, can transiently suppress phenotypic
accumulation of several mutations (Larder and resistance to ZDV in viruses that have ZDV resis-
Kemp, 1989). The level of ZDV resistance con- tance mutations (St Clair et al., 1991; Larder et
ferred by the K70R and T215Y/F mutations al., 1995). The molecular mechanisms of these
alone varies from 8– 16-fold. In contrast, combi- interactions are not well understood.
nation of these and other secondary mutations A small proportion ( 1%) of patients treated
such as K219Q or L210W decrease ZDV suscepti- with NRTIs develop resistance through a T69S
bility \100-fold (Kellam et al., 1994; Lacey and mutation and a family of insertion mutations
Larder, 1994a). Low level resistance to ddI and (usually two serines) between positions 69 and 70
ddC is associated with the L74V, K65R and of the RT. The presence of this insertion along
T69D mutations (St Clair et al., 1991; Kozal et with the T215Y mutation results in multiple
al., 1994; Zhang et al., 1994). The L74V, K65R, dideoxynucleoside resistance (MDNR) (Winters et
and M184V mutations also confer resistance to al., 1998; de Jong et al., 1999; Larder et al., 1999).
abacavir (Harrigan et al., 2000). The basis of d4T These findings illustrate the ability of HIV-1 to
resistance is not fully clear. Resistance to d4T has escape drug pressure even through amino acid
been associated with insertion mutations at codon insertions in a region of the enzyme that is critical
69, and the Q151M or V75T mutations (Lacey for the polymerase function (Huang et al., 1998).
and Larder, 1994b; Lin et al., 1999). However, Other mutations associated with MDNR are
d4T resistance occurs at low frequency and these A62V, V75I, F77L, F116Y, and Q151M (Shi-
mutations are rarely seen in patients who fail d4T rasaka et al., 1993, 1995; Ueno et al., 1995;
treatment (Lin et al., 1999). Iversen et al., 1996; Schmit et al., 1996; Garcı́a-
Lerma et al., 1999; Van Laethem et al., 2000).
Table 3
These mutations have been observed in 3–16% of
Mutations associated with resistance to HIV-1 protease in-
hibitors patients treated with ZDV and ddC/ddI, and con-
fer resistance to all currently approved nucleoside
Protease Primary Secondary mutations analogs including ZDV, ddI, ddC, d4T, 3TC, and
inhibitor mutations abacavir (Shafer et al., 1994, 1995; Schmit et al.,
Indinavir M46I, L10I/R/V, K20M/R, 1998; Kavlick et al., 1998). The emergence of
V82A/F/T/S L24I, V32I, M36I, these MDNR mutations compromises the clinical
I54V, A71V/T, efficacy of this entire class of compounds, and
G73S/A, V77I, I84V, therefore, severely limits treatment options.
L90M
Ritonavir V82A/F/T/S K20M/R, V32I, L33F,
Development of ZDV resistance in patients
M36I, M46I/L, treated with ZDV+ ddI/ddC is known to occur
I54V/L, A71V/T, V77I, through acquisition of either the T215Y or
I84V, L90M Q151M mutation but very rarely through both
Saquinavir G48V, L90M L10I/R/V, I54V,
mutation (Kavlick et al., 1998; Schmit et al.,
A71V/T, G73S, V77I,
V82A, I84V 1998). The viral determinants that may influence
Nelfinavir D30N, L90M Ll0F/I, M36I, M46I/L, selection of either Q151M or T215Y remain un-
A71V, V77I, defined. However, we have recently found indirect
V82A/F/T/S, I84V, evidence for a role of the viral background and an
N88D
Amprenavir I50V, I84V L10F/I/R/V, V32I,
intermediate mutation (Q151L) in the develop-
M46I, I47V, I54V ment of MDNR mediated by Q151M (Garcı́a-
Lerma et al., 2000a).
J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212 201

3.2. Resistance to NNRTIs Other mutations such as Y181C or P236L have


been observed at low frequency alone or in com-
The binding site of NNRTIs is a hydrophobic bination with K103N (Dueweke et al., 1993a;
pocket close to the polymerase catalytic site in the Demeter et al., 1997). The K103N mutation is
p66 subunit of RT. Since these compounds share also the mutation most frequently seen in patients
a common mechanism of action and bind to a who fail treatment with efavirenz. K103N alone
common site of the enzyme, any slight variation confers moderate resistance to efavirenz (20-fold
due to single point mutations may have a signifi- reduced susceptibility), and cross resistance to
cant impact on the sensitivity to this class of nevirapine and delavirdine. High level resistance
compounds. These point mutations are well char- to efavirenz requires combination of mutations
acterized and result in the loss of stabilizing inter- such as K103N/V108I, K103N/P225H, or
actions and the emergence of steric and K103N/L100I (Bacheler et al., 1999; Jeffrey et al.,
thermodynamic barriers for NNRTI binding 1999).
(Maga et al., 1997).
NNRTIs select for mutations in two different
regions of the RT (codons 98– 108 and codons 3.3. Resistance to protease inhibitors
179–190). Resistance to nevirapine is associated
with the A98G, L100I, K103N, V106A, V108I, To date, more than 20 amino acid substitutions
Y181C, Y188C, and G190A mutations (Richman have been observed in the HIV-1 protease follow-
et al., 1994; Havlir et al., 1995; D’Aquila et al., ing treatment with protease inhibitors (Table 2).
1996; Montaner et al., 1998). The Y181C muta- Primary mutations associated with resistance to
tion is the most frequently seen mutation in pa- protease inhibitors are relatively specific for each
tients treated with nevirapine monotherapy. This drug (e.g. L90M and G48V for saquinavir, or
mutation alone results in \ 100-fold decreased I50V and I84V for amprenavir). Cross resistance
susceptibility to nevirapine (Richman et al., 1991; among protease inhibitors is associated with the
Havlir et al., 1996). Selection of additional muta- presence of additional secondary mutations (Con-
tions associated with nevirapine resistance such as dra et al., 1995), which may appear later during
K103N, Y188C, and G190A is more frequently treatment or can pre-exist as natural polymor-
observed in patients receiving combination ther- phisms (Lech et al., 1996). However, the existence
apy with other drugs such as ZDV (Richman et of two or more primary mutations is likely to
al., 1994). confer broad cross-resistance to most currently
A recent clinical trial done in Uganda has available protease inhibitors (Condra et al., 1995;
shown that a single-dose of nevirapine monother- Lorenzi et al., 1999).
apy can reduce vertical HIV-1 transmission in
50% during the first 14 – 16 weeks of life compared
with the standard ZDV monotherapy, providing a
simple and inexpensive regimen that can be used 4. Resistance tests
in less-developed countries (Guay et al., 1999).
Preliminary evidence indicate that about one third Resistance of HIV-1 to RT or protease in-
of the women developed genotypic resistance to hibitors can be determined genotypically by exam-
nevirapine 6 weeks after treatment. However, the ining resistance-related mutations in the viral
mutation identified in all the women was K103N genome, and/or phenotypically by measuring the
and not the previously observed Y181C mutation inhibition of virus replication or enzymatic activ-
(Becker-Pergola et al., 2000). Selection of K103N ity of RT/protease by specific drugs. To date,
may probably be related to the non-subtype B several phenotypic and genotypic assays have
viral background. been developed and are being used to monitor for
The predominant mutation associated with drug resistance. The relative advantages of geno-
delavirdine resistance is the K103N mutation. typic and phenotypic assays are shown in Table 4.
202 J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212

Table 4
Advantages and disadvantages of phenotypic and genotypic assays

Assay Advantages Disadvantages

Phenotypic assays
Replication- Direct measure of drug susceptibility Expensive, laborious
based assays Can assess complex resistance patterns Long time to results (2–4 weeks)
Results are more familiar to clinicians Clinically relevant cutoff values not clearly defined

Enzymatic assays Rapid (1–2 days), simple, and cheap Rapid protease assays not developed
Can detect low proportion of resistant Currently unable to detect AZT resistance
viruses Useful for subtype B and non-subtype B HIV-1

Genotypic assays Cheaper and more rapid than replication- Indirect measure of susceptibility
based assays Requires expert clinical interprepation
Less technically demanding than replication- Resistance patterns not fully defined
based assays

4.1. Genotypic assays analysis provides information on all positions as-


sociated with drug resistance, and several compa-
All genotypic assays currently used are based nies are now developing kits including Visible
on PCR amplification of viral pol sequences pri- Genetics and ABI/Perkin Elmer.
marily from plasma by RT-PCR. Amplified prod- Expert clinical interpretation of genotypic re-
ucts can be analyzed by hybridization or sults is important and may often be complicated
sequencing techniques. Hybridization techniques by the presence of complex mutations patterns.
are illustrated by the commercially available Line Important issues for analysis of genotypic results
Probe Assay (LIPA HIV-1 RT, Innogenetics) that also include laboratory quality assurance, and use
detects key resistance mutations. The LIPA assay of appropriate controls. A recent study that used
is available as a kit, and is based on reverse well-defined coded samples has shown large inter
hybridization of biotin-labeled amplified HIV-1 laboratory differences in the quality of the results
sequences from plasma with short, immobilized obtained by automated DNA sequencing in 23
oligonucleotides. The hybrid is detected with a laboratories worldwide (Schuurman et al., 1999).
streptavidine-alkaline phosphatase-based colori- These findings highlight the need for proper qual-
metric procedure (Stuyver et al., 1997, 1999). The ity control and standardization among currently
assay detects both wild type and resistant vari- used sequencing methods.
ants, and has a high sensitivity in detecting low
proportion (10%) of resistant viruses (Stuyver 4.2. Phenotypic assays
et al., 1997). Fig. 1b shows the ability of the assay
to detect 10% of viruses carrying the M184V Phenotypic testing measures the ability of an
mutation in a background of wild type viruses. HIV-1 isolate to replicate in the presence of a
However, this method is available for genotyping drug or, alternatively, the enzymatic susceptibility
only selected RT mutations, and may occasionally of RT/protease to inhibition by a drug. Suscepti-
give false negative results due to the presence of bility of HIV-1 is determined by measuring the
polymorphisms/mutations near the site of the spe- drug concentrations that inhibit in 50 or 90%
cific hybridization (Kock et al., 1999; Puchham- virus replication (IC50 and IC90, respectively) or
mer-Stockl et al., 1999). In contrast, sequence enzymatic activity. Comparison of the IC50 and
J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212 203

IC90 values of a particular virus with those found evaluation with an expensive p24 sandwich
in a reference wild type HIV-1, provides a direct ELISA, is time consuming and labor intensive,
measurement of the level of resistance (i.e. fold and is also fraught with biologic variabilities,
increase in IC50) to the specific drug. including those related to virus isolation and
tropism.
To circumvent the problem of virus isolation
4.3. Replication-based assays
and cellular tropism, several recombinant virus
assays (RVA) have been developed. A schematic
Replication-based assays require the use of
HIV-1 clinical isolates. Conventionally, HIV-1 representation of the assay is shown in Fig. 2.
isolates are obtained from cultures of peripheral This assay uses patient-derived RT sequences that
blood lymphocytes or plasma. Drug susceptibility are amplified by PCR and an RT-deleted proviral
testing of these viral isolates can then be measured laboratory clone to generate recombinant viruses
by several methods including the HeLa CD4+ in a human T cell line by homologous recombina-
plaque reduction assay and the peripheral blood tion (Kellam and Larder, 1994). The first RVA
mononuclear cell (PBMC)-based culture system. was designed to measure susceptibility to RT
The HeLa CD4 + plaque reduction assay was inhibitors only, and used PBMC as a source of
the first standardized phenotypic test (Larder et viral sequences (Kellam and Larder, 1994). These
al., 1990). However, this assay is only suitable for recombinant viruses retain the drug susceptibility
syncytium-inducing (SI) strains, and most patients of the RT present in the clinical specimen and can
isolates contain nonsyncytium-inducing (NSI) be cultured in vitro without the selective effect of
strains or a mixture of SI and NSI strains. The virus tropism. The assay was later modified to use
PBMC-based culture system, in contrast, could be plasma viral RNA, and to allow drug susceptibil-
used in more than 80% of clinical isolates. A ity testing to protease inhibitors (Hertogs et al.,
standardized protocol for drug susceptibility test- 1998).
ing in PBMCs, called the ACTG-DoD (AIDS To date, two RVA are commercially available
Clinical Trials Group, Department of Defense) is to measure susceptibility to RT and protease in-
widely used for drug susceptibility testing (Japour hibitors: Virco Antivirogram™ and ViroLogic
et al., 1993). However, this assay requires a final PhenoSense™. Both assays amplify HIV-1 RT

Fig. 1. Detection threshold of 3TC resistance mediated by the M184V mutation by a phenotypic (Amp-RT) or a genotypic (LIPA)
assay (adapted from Garcı́a-Lerma et al., 1999). (A) Levels of RT inhibition by 3TC-TP measured by Amp-RT in mixtures of wild
type and 3TC-resistant viruses; (B) genotypic detection of the M184V mutation by the HIV-1 LIPA in the same virus mixtures.
204 J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212

The Virco Antivirogram™ assay (Antiviro-


gram; Virco, Mechelen, Belgium) is based on in-
tracellular homologous recombination of
patient-derived gag/RT/protease sequences and a
gag/RT/protease-deleted proviral clone. Replica-
tion of the recombinant viruses is analyzed in the
presence of various concentrations of antiretrovi-
ral drugs using a reporter gene-based cellular
assay (Pauwels et al., 1998).
A comparative analysis of Virco Antiviro-
gram™ and ViroLogic Phenosense™ has been
recently done using coded plasma samples from
drug-naı̈ve or drug-experience HIV-1-infected per-
sons (Qari et al., 2000). The results showed a 92%
concordance between results by both assays, with
the majority of the discordant results observed
among samples that had IC50 values close to assay
cut-off values. These findings indicate that despite
Fig. 2. Schematic representation of the first recombinant virus the use of different testing strategies in both as-
assay developed by Kellam and Larder (1994). The assay
says, there is a good correlation between test
allowed the generation of viable viruses by homologous re-
combination of a PCR-derived pool of RT coding sequences results.
from a patient and an RTdeleted, noninfectious proviral clone. While phenotypic data are easy to interpret in
IC50 and IC90 values for drug were determined using a T cell both assays, there are at present no well defined,
line. clinically validated cut-off values that correlate
virologic response with IC50 values. It may be
and protease but differ in technical aspects related reasonable to expect that the higher the level of
to recombinant virus construction and detection resistance in vitro, the less activity the drug will
of virus replication. Drug susceptibility results are have in vivo. However, low level resistance can
reported as IC50 values for each drug and fold also be associated with a poor response.
differences of these values relative to a reference
wild type virus. Fold differences \4 and \2.5 4.4. Enzymatic-based assays for phenotypic RT
are considered evidence of decreased susceptibility susceptibility testing
for the Antivirogram and PhenoSense assays,
RT and protease assays have been widely used
respectively.
as research tools to measure drug susceptibility
The ViroLogic PhenoSense™ assay (ViroLogic,
(Ueno et al., 1995; Furline, 1999). However, the
South San Francisco, CA) measure susceptibility
use of these assays for drug susceptibility testing
by using resistance test vectors (RTVs) that con-
in plasma has not been possible because of the
tain a luciferase indicator gene and patient- low sensitivity of these assays. Recently, several
derived RT and protease sequences (Petropoulos ultrasensitive RT assays such as PERT and Amp-
et al., 2000). Basically, cells transfected with RT have been described (Silver et al., 1993; Pyra
RTVs produce viral particles that are used to et al., 1994; Heneine et al., 1995; Boni et al.,
infect target cells. Since RTVs are replication 1996). All these assays use PCR amplification for
defective, luciferase activity is measured after a the detection of the RT-generated cDNA but
single round of viral replication (Petropoulos et differ in the RNA template and some testing
al., 2000). Luciferase activity measured in the conditions. We have developed Amp-RT-based
presence of RT and protease inhibitors is used to assays for the analysis of RT susceptibility to
measure the level of drug resistance. RTIs and NNRTIs. The Amp-RT assay detects
J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212 205

RT activity by using a known heterologous RNA reactions done in the presence of drug, while no
template derived from the encephalomyocarditis detectable RT signal is seen in RTs from sensitive
virus (EMCV) RNA genome. The RT-derived isolates.
EMCV cDNA is detected by PCR amplification Susceptibility testing of HIV-1 RT by Amp-RT
and an ELISA-based hybridization with an inter- is done by measuring IC50 and IC90 of the RT
nal EMCV-specific probe (Heneine et al., 1995; enzyme to NNRTIS or the triphosphate form of
Yamamoto et al., 1996; Garcı́a-Lerma et al., NRTIs. Amp-RT IC50 values determined in sev-
1998). Phenotypic resistance to RT inhibitors is eral wild type and nevirapine- or 3TC-resistant
measured by quantitating the RT signal generated isolates have been shown to correlate with those
in Amp-RT reactions done in the presence and measured by culture-based phenotypic assays
absence of the relevant RT inhibitor. Fig. 3 illus- (Garcı́a-Lerma et al., 1999; Vázquez-Rosales et
trates the principle of this testing approach. For al., 1999). In contrast to conventional phenotypic
resistant RTs, an Amp-RT signal is seen in RT assays, drug susceptibility testing by Amp-RT

Fig. 3. Principle of the Amp-RT-based phenotypic assay for detection of resistance to RT inhibitors. Amp-RT measures the ability
of HIV-1 RT to produce a cDNA copy from an encephalomyocarditis (EMCV) RNA template. HIV-1 RT from plasma is tested
in the absence and presence of several concentrations of drug. The RT-generated EMCV cDNA is detected by PCR amplification
and quantitated by ELISA. IC50 and IC90 for drug are determined and compared with a wild type virus reference virus.
206 J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212

IC50 for nevirapine (Fig. 4). A similar screening


assay was also successfully used for detecting
phenotypic resistance to 3TC in plasma mediated
by the M184V or the MDNR mutations (Garcı́a-
Lerma et al., 1999) (Fig. 5). Both assays have the
ability to detect low proportions ( 10%) of resis-
tant viruses, and therefore, may be used for early
detection of resistance to these drugs (Garcı́a-
Lerma et al., 1999; Vázquez-Rosales et al., 1999)
(Fig. 1a).
The mechanisms of ZDV resistance are not
completely understood and affect the use of the
Amp-RT assay to measure ZDV resistance. RTs
carrying the classical ZDV resistance mutations
(i.e. M41L, T215Y/F, K70R or D67N) have Ki
values or IC50 values for ZDV-TP that are similar
to wild type RTs (Arts and Wainberg, 1996). This
Fig. 4. Detection of phenotypic resistance to nevirapine in
plasma by Amp-RT (adapted from Vázquez-Rosales et al.,
unique enzymatic characteristic precludes the use
1999). (A) Level of RT inhibition by 50 mM nevirapine and of RT assays to detect ZDV resistance. However,
correlation with detection of the Y181C mutation by differen- a recent report has shown that efficient discrimi-
tial hybridization. Open circles correspond to the RT inhibi- nation between wild type and ZDV-resistant RTs
tion values determined by Amp-RT, and closed circles can be achieved enzymatically by adding ATP or
correspond to the log10 ratio between mutant genotypes and a
pyrophosphate to the RT reaction (Lennerstrand
highly conserved wild type region of the HIV-1 RT (MUT/
GNR), (B) Amp-RT IC50 values for nevirapine in sequential et al., 2000). Therefore, using these modified assay
samples from a patient treated with nevirapine. Sequential conditions it may be possible to test enzymatically
samples from days 6, 7, 21, and 28 were tested in the absence for ZDV resistance.
and presence of several concentrations of nevirapine, and Amp-RT-based phenotypic assays provide a
inhibition of RT activity was computed.
generic tool that can also be used to monitor for
drug resistance in persons infected with highly
does not require virus isolation and culture and, divergent viruses. We have found that the re-
therefore, provides rapid information (1– 2 days) bound in RT-based plasma virus load observed in
on resistance. two HIV-1 group O-infected patients treated with
A simple testing strategy using one drug con- d4T, 3TC and indinavir, was associated with evi-
centration in the Amp-RT assay was also found dence of phenotypic resistance to Lamivudine by
to be useful for rapid screening of resistance in Amp-RT and with the detection of the M184V
plasma samples. We have demonstrated the valid- mutation. These Amp-RT-based phenotypic as-
ity of this testing approach for detecting resistance says should therefore facilitate studies aimed at
to 3TC and nevirapine (Garcı́a-Lerma et al., 1999; evaluating the responses of HIV-1 group O-in-
Vázquez-Rosales et al., 1999). Fig. 4 compares fected patients to RT and protease inhibitors
Amp-RT-based phenotypic results with detection (Garcı́a-Lerma et al., 2000b).
of the Y181C mutation in patients treated with
nevirapine. A correlation between RT susceptibil-
ity by Amp-RT and the levels of Y181C mutation 5. Use of resistance testing for clinical
was found in longitudinally samples collected management
from patients receiving nevirapine monotherapy
(Vázquez-Rosales et al., 1999). The decrease in Data from various retrospective and prospec-
the level of RT inhibition observed overtime was tive studies are accumulating to support the use of
also associated with an increase in the Amp-RT drug resistance testing in selecting drugs in many
J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212 207

Fig. 5. Detection of phenotypic resistance to 3TC by analysis of RT inhibition by 5 uM 3TC-TP in the Amp-RT assay. Amp-RT
reactions done in the presence and absence of 3TC-TP are shown. Lanes 1 and 2, wild type (wt) HIV-1 RT; Lane 3, 3TC-resistant
(M184V); Lane 4, nevirapine-resistant (181C/Y); lane 5, 3TC/nevirapine-resistant (M184V/YI81C); Lane 6, AZT-resistant (D67N/
K70R/T215F/K219Q); Lanes 7, 8, and 9, HIV-1 RT carrying different multidrug-resistant mutations; Neg, uninfected culture
supernatant.

clinical situations (summarized in Hirsch et al., References


2000).
Two recent prospective randomized trials have Arts EJ, Wainberg MA. Mechanisms of nucleoside analog
examined the benefits of genotype testing in pa- antiviral activity and resistance during human immunodefi-
ciency virus reverse transcription. Antimicrob Agents
tient management. The VIRADAPT study found
Chemother 1996;40:527 – 40.
a significantly better virologic response after 6 Bacheler LT, Anton B, Baker D, Mecker M, Lasut A, Aujay
months of treatment in a group of patients who M, Bolling L, Krakowski K, Bunville J, Wang V, Abrem-
had their therapy changed according to genotype sky K. Genotypic correlates of in vivo resistance to efa-
testing results compared to usual standard of care virenz. 6th Conference on Retroviruses and Opportunistic
(Durant et al., 1999). The benefit of resistance Infections 1999. Abstr. 109, p. 88.
Baxter JD, Mayers DL, Wentworth DN, Neaton JD, Merigan
testing was maintained for 1 year (Clevenbergh et TC, and the CPCRA 046 Study Team for the Terry Beirn
al., 1999). The genotyping antiviral resistance test- Community Programs for Clinical Research on AIDS
ing (GART) study found a significantly better (CpcRA). Final results of CPCRA 046: a pilot study of
virologic response after 8 weeks with treatment antiretroviral management based on plasma genotypic an-
regimen based on genotype and expert advise tiretroviral resistance testing (GART) in patients failing
antiretroviral therapy. Antiviral Ther 1999;4(Suppl 1):43
versus standard of care (Baxter et al., 1999).
Becker-Pergola G, Guay L, Miro F, Musoke P, Fung S,
An international AIDS Society-USA panel re- Jackson JB, Eshleman SH. Selection of the K103N nevi-
cently recommended drug resistance testing to rapine (NVP) resistance mutation in Ugandan women
help guide the choice of new regimens after treat- receiving NVP prophylaxis to prevent HIV-1 vertical trans-
ment failure and for guiding therapy for HIV- mission (HIVNIET-006). In: 7th Conference on Retro-
infected pregnant women. The panel also sug- viruses and Opportunistic Infections 2000. Abstr. 658, 198.
Boni J, Pyra H, Schupbach J. Sensitive detection and quantifi-
gested resistance testing to be considered in treat-
cation of particle-associated reverse transcriptase in plasma
ment-naive patients with established infection or of HIV-1-infected individuals by the product-enhanced
prior to initiating therapy in patients with acute reverse transcriptase (PERT) assay. J Med Virol
HIV infection (Hirsch et al., 2000). However, 1996;49:23 – 8.
drug resistance test results should not be used as Boucher CAB, Cammack N, Scipper P, Schuurman R, Rouse
the principal criteria for decisions on initiating or P, Wainberg MA, Cameron JM. High level resistance to
( −) enantiomeric 2-deoxy-3%-thiacytidine in vitro is due to
changing antiretroviral therapy. Such decisions one amino acid substitution in the catalytic site of human
should be based primarily on plasma virus loads immunodeficiency virus type 1 reverse transcriptase.
(Hirsch et al., 2000) Antmicrob Agents Chemother 1993;37:2231 – 4.
208 J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212

Clevenbergh P, Durant J, Halfon P, et al. Persisting long-term Domingo E, Baranowski E, Ruiz-Jarabo CM, Martin-Hernán-
benefit of antiretroviral genotypic guided treatment for dez AM, Saiz JC, Escarmis C. Quasispecies structure and
HIV-infected patients failing on HAART: the viraclapt persistance of RNA viruses. Emerg Infect Dis 1998;4:521 –
study, week 48 follow up. Antiviral Ther 1999;4(suppl 7.
1):42. Domingo E, Holland JJ, Biebricher C, Eigen M. Quasispecies:
Coates JA, Cammack N, Jenkinson HJ, Jowett AJ, Jowett MI, the concept and the word. In: Gibbs A, Calisher C, Garcia-
Pearson BA, Penn CR, Rouse PL, Viner KC, Cameron Arenal F, editors. Molecular Basis of Virus Evolution.
JM. 2%-deoxy-3%-thiacytidine is a potent, highly selective Cambridge: Cambridge University Press, 1995:171 – 80.
inhibitor of human immunodeficiency virus type 1 and type Domingo E, Menéndez-Arias L, Quiñones-Mateu ME, Hol-
2 replication in vitro. Antimicrob Agents Chemother guı́n A, Gutierrez-Rivas M, Martinez MA, Quer J, Novella
1992;36:733 – 9. IS, Holland JJ. Viral quasispecies and the problem of
Condra JH, Schleif WA, Blahy OM, Gabryelski LJ, Graham vaccine-escape and drug-resistant mutants. In: Juker E,
DJ, Quintero JC, Rhodes A, Robbins HL, Roth E, Shiv- editor. Progress in Drug Research. Basel: Birkhauser,
aprakash M, Titus D, Yang T, Teppler H, Squires KE, 1997:99 – 128.
Deutsch PJ, Emini EA. In vivo emergence of HIV-1 vari- Dueweke TJ, Pushkarskaya T, Poppe SM, Swaney SM, Zhao
ants resistant to multiple protease inhibitors. Nature JQ, Chen IS, Stevenson M, Tarpley WG. A mutation in
1995;374:569 – 71. reverse transcriptase of bis (heteroaryl) piperazineresistant
Daluge SM, Good SS, Faletto MB, Miller MH, St Clair MH, human immunodeficiency virus type 1 that confers in-
Boone LR, Tisdale M, Parry NR, Reardon JE, Dornsife creased sensitivity to other nonnucleoside inhibitors. Proc
RE, Averett DR, Krenitsky TA. 1592U89, a novel carbo- Natl Acad Sci USA 1993a;90:4713 – 7.
cyclic nucleoside analog with potent, selective anti-human Dueweke TJ, Poppe SM, Romero DL, Swaney SM, So AG,
immunodeficiency virus activity. Antimicrob Agents Downey KM, Althaus IW, Reusser F, Busso M, Resnick
Chemother 1997;41:1082 –93. L, et al. U-90152, a potent inhibitor of human im-
D’Aquila RT, Hughes MD, Johnson VA, Fischl MA, Som- munodeficiency virus type 1 replication. Antimicrob
madossi JP, Liou SH, Timpone J, Myers M, Basgoz N, Agents Chemother 1993b;37:1127 – 31.
Niu M, Hirsch MS. Nevirapine, zidovudine, and di- Durant JCP, Clevenbergh P, Halfon P, Delgiudice P, Porsin S,
danosine compared with zidovudine and didanosine in Simonet P, Montagne N, Boucher CA, Schapiro JM, Del-
patients with HIV-1 infection. A randomized, double- lamonica P. Drug-resistance genotyping in HIV-1 therapy:
blind, placebo-controlled trial. National Institute of Al- the VIRADAPT randomized controlled trial. Lancet
lergy and Infectious Diseases AIDS Clinical Trials Group 1999;353:2195 – 9.
Protocol 241 Investigators. Ann Intern Med Furline ES. Identifying and characterizing HIV protease in-
1996;124:1019 –30. hibitors. In: Kinchington D, Schinazi RF, editors. Meth-
de Jong JJ, Goudsmit J, Lukashov V, Hillebrand ME, Huis- ods in Molecular Medicine: Antiviral Methods and
mans R, Danner SA, ten Veen JH, de Wolf F, Jurriaans S. Protocols, vol. 24. Totowa, NJ: Humana Press, 1999:313 –
Insertion of two amino acids combined with changes in 25.
reverse transcriptase containing tyrosine-215 of HIV-1 re- Garcı́a-Lerma JG, Yamamoto S, Gómez-Cano M, Soriano V,
sistant to multiple nucleoside analogs. AIDS 1999;13:75 – Green TA, Busch MP, Folks TM, Heneine W. Measure-
80. ment of human immunodeficiency virus type 1 plasma
de Jong MD, Schuurrman R, Lange JMA, Boucher CAB. virus load based on reverse transcriptase (RT) activity:
Replication of a pre-existing resistant HIV-1 sub popula- evidence of variabilities in levels of virion-associated RT. J
tion in vivo after introduction of a strong selective drug Infect Dis 1998;177:1221 – 9.
pressure. Antiviral Ther 1996;1:33 – 41. Garcı́a-Lerma JG, Schinazi RF, Juodawlkis AS, Soriano V,
Demeter LM, Meehan PM, Morse G, Gerondelis P, Dexter A, Lin Y, Tatti K, Rimland D, Folks TM, Heneine W. A
Berrios L, Cox S, Freimuth W, Reichman RC. HIV-1 drug rapid non-culture-based assay for clinical monitoring of
susceptibilities and reverse transcriptase mutations in pa- phenotypic resistance of human immunodeficiency virus
tients receiving combination therapy with didanosine and type 1 to lamivudine (3TC). Antimicrob Agents Chemother
delavirdine. J AIDS 1997;14:136 –44. 1999;43:264 – 70.
Descamps D, Colling G, Loussert-Ajaka I, Saragosti S, Simon Garcı́a-Lerma JG, Gerrish P, Wright AJ, Qari SH, Heneine
F, Brun-Vezinet F. HIV-1 group O sensitivity to antiretro- W. Evidence of a role for the Q151L mutation and the
viral drugs. Acquir Immune Defic Syndr Hum Retrovirol viral background in the development of multiple
1995;9:977 – 8. dideoxynucleoside resistant HIV-1, J. Virol 2000;74:9339 –
Descamps D, Collin G, Letourneur F, Apetrei C, Damond F, 46.
Loussert-Ajaka I, Simon F, Saragosti S, Brun-Vezinet Garcı́a-Lerma JG, Soriano V, Mas A, Quiñones-Mateu ME,
F. Susceptibility of human immunodeficiency virus Arts EJ, Heneine W. Quantitation of human immunodefi-
type 1 group O isolates to antiretroviral agents: in vitro ciency virus type 1 group O in plasma by measuring
phenotypic and genotypic analysis. J Virol 1997;71:8893 – reverse transcriptase activity. J Clin Microbiol
8. 2000b;38:402 – 5.
J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212 209

Guay LA, Musoke P, Fleming T, Bagenda D, Allen M, Cauwenberge A, Van den Eynde C, Van Gerwen V, Azijn
Nakabiito C, Sherman J, Bakaki P, Ducar C, Deseyve M, H, Van Houtte M, Peeters F, Staszewski S, Conant M,
Emel L, Mirochnick M, Gleen Fowler M, Mofenson L, Bloor S, Kemp S, Larder B, Pauwels R. A rapid method
Miotti P, Dransfield K, Bray D, Mmiro F, Jackson J. for simultaneous detection of phenotypic resistance to in-
Intrapartum and neonatal single-dose nevirapine compared hibitors of protease and reverse transcriptase in recombi-
with zidovudine for prevention of mother-to-child trans- nant human immunodeficiency virus type 1 isolates from
mission of HI-1 in Kampala, Uganda: HIVNET 012 ran- patients treated with antiretroviral drugs. Antimicrob
domized trial. Lancet 1999;3:795 –802. Agents Chemother 1998;42:269 – 76.
Gulick RM, Mellors JW, Havlir D, Eron JJ, Gonzalez C, Hirsch MS, Brun-Vezinet F, D’Aquila RT, Hammer SM,
McMahon D, Richman DD, Valentie FT, Jonas L, Mei- Johnson VA, Kuritzkes DR, Loveday C, Mellors JW,
bohm A, Emini EA, Chodakewitz JA. Treatment with Clotet B, Conway B, Demeter LM, Vella S, Jacobsen DM,
indinavir, zidovudine, and lamivudine in adults with hu- Richman DD. Antiretroviral drug resistance testing in
man immunodeficiency virus infection and prior antiretro- adult HIV-1 infection: recommendations of an interna-
viral therapy. New Engl J Med 1997;337:734 –9. tional AIDS society-USA panel. J Am Med Assoc
Hammer SM, Squires KE, Hughes MD, Grimes JM, Demeter 2000;283:2417 – 26.
LM, Currier JS, Eron JJ, Feinberg JE, Balfour HH, Dey- Ho DD, Toyoshima T, Mo H, Kempf DJ, Norbeck D, Chen
ton LF, Chodakewitz JA, Fischl MA. AIDS Clinical Trials CM, Wideburg NE, Burt SK, Erickson JW, Singh MK.
Group 320 Study Team. A controlled trial of two nu- Characterization of human immunodeficiency virus type 1
cleoside analogues plus indinavir in persons with human variants with increased resistance to C2-symmetric
immunodeficiency virus infection and CD4 cell counts of protease inhibitor. J Virol 1994;68:2016 – 20.
200 per cubic milliliter or less. New Engl J Med Hooker DJ, Tachedjian G, Solomon AE, Gurusinghe AD,
1997;337:725 – 33. Land S, Birch C, Anderson JL, Roy BM, Arnold E,
Harrigan PR, Kinghorn I, Bloor S, Kemp SD, Najera I, Kohli
Deacon NJ. An in vivo mutation from leucine to tryp-
A, Larder BA. Significance of aminoacid variation of
tophan at position 210 in human immunodeficiency virus
human immunodeficiency virus type 1 reverse transcriptase
type 1 reverse transcriptase contributes to high-level resis-
residue 210 for zidovudine susceptibility. J Virol
tance to 3%-azido-3%-deoxythymidine. J Virol 1996;70:8010 –
1996;70:5930 – 4.
8.
Harrigan PR, Stone C, Griffin P, Najera I, Bloor S, Kemp S,
Huang H, Chopra R, Verdine L, Harrison SC. Structure of a
Tisdale M, Larder B. Resistance profile of the human
covalently trapped catalytic complex of HIV-1 reverse
immunodeficiency virus type 1 reverse transcriptase in-
transcriptase: implications for drug resistance. Science
hibitor abacavir (1592U89) after monotherapy and combi-
1998;282:1669 – 775.
nation therapy. CNA2001 Investigative Group. J Infect
Iversen AK, Shafer RW, Wehrly K, Winters MA, Mullins JI,
Dis 2000;181:912 – 20.
Chesebro B, Merigan TC. Multidrug-resistant human im-
Havlir D, Eastman S, Gamst A, Richman DD. Nevirapine-re-
sistant human immunodeficiency virus: kinetics of replica- munodeficiency virus type 1 strains resulting from combi-
tion and estimated prevalence in untreated patients. J Virol nation antiretroviral therapy. J Virol 1996;70:1086 – 90.
1996;70:7894 – 9. Japour AJ, Mayers DL, Johnson VA, Kuritzkes DR, Beckett
Havlir D, McLaughlin M, Richman DD. A pilot study to LA, Arduino JM, Lane J, Black RJ, Reichelderfer PS,
evaluate the development of resistance to nevirapine in D’Aquila RT, et al. Standardized peripheral blood
asymptomatic human immunodeficiency virus-infected pa- mononuclear cell culture assay for determination of drug
tients with CD4 cell counts \ 500/mm3: AIDS Clinical susceptibilities of clinical human immunodeficiency virus
Trial Group Protocol 208. J Infect Dis 1995;172:1379 – type 1 isolates. Antimicrob Agents Chemother
83. 1993;37:1095 – 101.
Heneine W, Yamamoto S, Switzer WM, Spira TJ, Folks TM. Jeffrey S, Corbett J, Bacheler L. In vitro NNRTI resistance of
Detection of reverse transcriptase by a highly sensitive recombinant HIV carrying mutations observed in efavirenz
assay in sera from persons infected with human im- treatment failures. In: 6th Conference on Retroviruses and
munodeficiency virus type 1. J Infect Dis 1995;171:1210 – Opportunistic Infections, 1999, Abstract 110, 88.
6. Joly V, Yeni P. Non nucleoside reverse transcriptase in-
Hertogs K, Bloor S, De Vroey V, van Den Eynde C, De- hibitors. AIDS Rev 1999;1:3744.
hertogh P, van Cauwenberge A, Sturmer M, Alcorn T, Kaldor SW, Kalish VJ, Davies JF, Shetty BV, Fritz JE, Appelt
Wegner S, van Houtte M, Miller V, Larder BA. A novel K, Burgess JA, Campanale KM, Chirgadze NY, Clawson
human immunodeficiency virus type 1 reverse transcriptase DK, Dressman BA, Hatch SD, Khalil DA, Kosa MEB,
mutational pattern confers phenotypic lamivudine resis- Lubbehusen PP, Muesing MA, Patick AK, Reich SH, Su
tance in the absence of mutation 184V. Antimicrob Agents KS, Tatlock JH. Viracept (nelfinavir mesylate, AG1343): a
Chemother 2000;44:568 – 73. potent orally bioavailable inhibitor of HIV-1 protease. J
Hertogs KM, de Bethune M, Miller V, Ivens T, Schel P, van Med Chem 1997;40:3979 – 85.
210 J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212

Kellam P, Larder BA. Recombinant virus assay: a rapid, Lech WJ, Wang G, Yang YL, Chee Y, Dorman K, McCrae
phenotypic assay for assessment of drug susceptibility of D, Lazzeroni LC, Erickson JW, Sinsheimer JS, Kaplan
human immunodeficiency virus type 1 isolates. Antimicrob AH. In vivo sequence diversity of the protease of human
Agents Chemother 1994;38:23 –30. immunodeficiency virus type 1: presence of protease in-
Kellam P, Boucher CA, Tijnagel JM, Larder BA. Zidovudine hibitor-resistant variants in untreated subjects. J Virol
treatment results in the selection of human immunodefi- 1996;70:2038 – 43.
ciency virus type 1 variants whose genotypes confer in- Lennerstrand J, Hertogs K, Starnmers D. Mechanisms of AZT
creasing levels of drug resistance. J Gen Virol resistance resolved biochemically: study of a panel of site-
1994;75:341 – 51. directed HIV-1 reverse transcriptase mutants. In: 7th Con-
Kock N, Yahi N, Colson P, Fantini J, Tamalet C. Genetic ference on Retroviruses and Opportunistic Infections,
polymorphisms near HIV-1 reverse transcriptase resis- 2000; abstract 734, 212.
tance-associated codons is a major obstacle for the line Lin PF, Gonzalez CJ, Griffith B, Friedland G, Calvez V,
probe assay as an alternative method to sequence analysis. Ferchal F, Schinazi RF, Shepp DH, Ashraf AB, Wainberg
J Virol Methods 1999;80:25 –31. MA, Soriano V, Mellors JW, Colonno RJ. Stavudine
Kohl NE, Emini E, Schleif WA, Davis LJ, Heimbach JC, resistance: an update on susceptibility following prolonged
Dixon RA, Scolnik EM, Sigal IS. Active human im- therapy. Antiviral Ther 1999;4:21 – 8.
munodeficiency virus protease is required for viral infectiv- Lin TS, Schinazi RF, Prusoff WH. Potent and selective in
ity. Proc Natl Acad Sci USA 1988;85:4686 –90. vitro activity of 3%-deoxythymidin-2%-ene (3%-deoxy-2%,3%-
Kavlick MF, Wyvill K, Yarchoan R, Mitsuya H. Emergence didehydrothymidine) against human immunodeficiency
of multi-dideoxynucleoside-resistant human immunodefi- virus. Biochem Pharmacol 1987;36:2713 – 8.
ciency virus type 1 variants, viral sequence variation, and Lorenzi P, Opravil M, Hirschel B, Chave JP, Furrer HJ, Sax
disease progression in patients receiving antiretroviral H, Perneger TV, Perrin L, Kaiser L, Yerly S. Impact of
chemotherapy. J Infect Dis 1998;98:1506 –13. drug resistance mutations on virologic response to salvage
Kosalaraksa P, Kavlick MF, Maroun V, Le F, Mitsuya H.
therapy. AIDS 1999;13:F17 – 21.
Comparative fitness of multidideoxynucleoside-resistant
Maeda Y, Venzon DJ, Mitsuya H. Altered drug sensitivity,
human immunodeficiency virus type 1 (HIV-1) in an in
fitness, and evolution of human immunodeficiency virus
vitro competitive HIV-1 replication assay. J Virol
type 1 with pol gene mutations conferring multidideoxynu-
1999;73:5356 – 63.
cleoside resistance. J Infect Dis 1998;177:1207 – 13.
Kozal MJ, Kroodsma K, Winters MA, Shafer RW, Efron B,
Maga G, Amacker M, Ruel N, Hubscher U, Spadari S.
Katzenstein DA, Merigan TC. Didanosine resistance in
Resistance to nevirapine of HIV-1 reverse transcriptase
HIV-infected patients switched from zidovudine to di-
mutants: loss of stabilizing interactions and thermody-
danosine monotherapy. Ann Intern Med 1994;121:263 –8.
namic or steric barriers are induced by different single
Lacey SF, Larder BA. Mutagenic study of codons 74 and 215
amino acid substitutions. J Mol Biol 1997;274:738 – 47.
of the human immunodeficiency virus type 1 reverse tran-
Mansky LM, Temin HM. Lower in vivo mutation rate of
scriptase, which are significant in nucleoside analog resis-
tance. J Virol 1994a;68:3421 –4. human immunodeficiency virus type 1 than that predicted
Lacey SF, Larder BA. Novel mutation (V75T) in human from the fidelity of purified reverse transcriptase. J Virol
immunodeficiency virus type 1 reverse transcriptase confers 1995;69:5087 – 94.
resistance to 2%,3%-didehydro-2%,3%-dideoxythymidine in cell Martı́nez-Picado J, Savara AV, Sutton L, D’Aquila RT.
culture. Antimicrob Agents Chemother 1994b;38:1428 – 32. Replicative fitness of protease inhibitor-resistant mutants
Larder BA, Chesebro B, Richman DD. Susceptibilities of of human immunodeficiency virus type 1. J Virol
zidovudine-susceptible and-resistant human immunodefi- 1999;73:3744 – 52.
ciency virus isolates to antiviral agents by using a quantita- Mitsuya H, Broder S. Inhibition of the in vitro infectivity and
tive plaque reduction assay. Antimicrob Agents Chemother cytopathic effect of human T-lymphotrophic virus type
1990;34:436 – 41. III/lymphadenopathy-associated virus (HTLV-HI/LAV)
Larder BA, Kemp SO. Multiple mutations in HIV-1 reverse by 2%,3%-dideoxynucleosides. Proc Natl Acad Sci USA
transcriptase confer highlevel resistance to zidovudine 1986;83:1911 – 5.
(AZT). Science 1989;246:1155 –7. Mitsuya H, Weinhold KJ, Furman PA, St Clair ME, Lehrman
Larder BA, Kemp SO, Harrigan PR. Potential mechanism for SN, Gallo RC, Bolognesi D, Barry DW, Broder S. 3%-
sustained antiretroviral efficacy of AZT+ 3TC combina- azido-3%-deoxythymidine (BW A509U): and antiviral agent
tion therapy. Science 1995;269:696 –9. that inhibits the infectivity and cytopathic effect of human
Larder BA, Bloor S, Kemp SD, Hertogs K, Deesmet RL, T-lymphotropic virus type III/lymphadenopathy-associated
Miller V, Sturmer M, Staszewski S, Ren J, Starnmers K, virus in vitro. Proc Natl Acad Sci USA 1985;82:7096 –
Stuart DI, Pauwels R. A family of insertion mutations 8100.
between codons 67 and 70 of human immunodeficiency Montaner JS, Reiss P, Cooper D, Vella S, Harris M, Conway
virus type 1 reverse transcriptase confer multinucleoside B, Wainberg MA, Smith D, Robinson P, Hall D, Myers
analog resistance. Antimicrob Agents Chemother M, Lange JM. A randomized, double-blind trial compar-
1999;43:1961 – 7. ing combinations of nevirapine, didanosine, and zi-
J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212 211

dovudine for HIV-infected patients. The INCAS trial. J Pauletti D. Nevirapine resistance mutations of human im-
Am Med Assoc 1998;279:930 –7. munodeficiency virus type 1 selected during therapy. J
Nájera I, Richman DD, Olivares I, Rojas JM, Peinado MA, Virol 1994;68:1660 – 6.
Perucho M, Nájera R, López-Galı́ndez C. Natural occur- Richman D, Shih CK, Lowy I, Rose J, Prodanovich P, Goff S,
rence of drug resistance mutations in the reverse transcrip- Griffin J. Human immunodeficiency virus type 1 mutants
tase of human immunodeficiency virus type 1 isolates. to nonnucleoside inhibitors of reverse transcriptase arise in
AIDS Res Hum Retrovir 1994;10:1479 –88. tissue culture. Proc Natl Acad Sci USA 1991;88:11241 – 5.
Nájera I, Holguı́n A, Quiñones-Mateu ME, Muñoz-Fernández Roberts NA, Martin JA, Kinchington D, Broadhurst AV,
MA, Nájera R, López-Galı́ndez C, Domingo E. Pol gene Craig JC, Duncan IB, Galpin SA, Handa BK, Kay J,
quasispecies of human immunodeficiency virus: mutations Krohn A, Lambert AW, Merret JH, Mills JS, Parkers
associated with drug resistance in virus from patients unde- KEB, Redshaw S, Ritchie AJ, Taylor DL, Thomas GJ,
going no drug therapy. J Virol 1995;69:23 –31. Machin PJ. Rational design of peptidebased HIV
Parteledis JA, Yamaguchi K, Tisdale M, Blair EE, Falcione C, proteinase inhibitors. Science 1990;248:358 – 61.
Maschera B, Myers RE, Pazhanisamy S, Futer O, Cullinan Schinazi RF, Lloyd RM, Nguyen MH, Cannon DL, McMil-
AB. In vitro selection and characterization of human im- lan A, Ilksoy N, Chu CK, Liotta DC, Bazmi HZ, Mellors
munodeficiency virus type 1 (HIV-1) isolates with reduced W. Characterization of human immunodeficiency viruses
sensitivity to hydroxyethylarnino sulfonamide inhibitors of resistant to oxatiolate-cytosine nucleosides. Antimicrob
HIV-1 aspartyl protease. J Virol 1995;69:5228 – 35. Agents Chemother 1993;37:875 – 81.
Pauwels R, Hertogs K, Kemp S, Bloor S, Van Acker K, Schmit J-C, Cogniaux J, Hermans P, Van Vaeck C, Sprecher
Hansen J, De Beukeleer W, Roeland C, Larder B, Stoffels S, Van Remoortel B, Witvrouw M, Blazarini J, Desmyter
P. Comprehensive HIV drug resistance monitoring using J, De Clercq E, Vandamme A-M. Multiple drug resistance
rapid, high-throughput phenotypic and genotypic assays to nucleoside analogs and nonnucleoside reverse transcrip-
with correlative data analysis. In: 2nd International Work- tase inhibitors in an efficiently replicating human im-
shop on HIV Drug Resistance and Treatment Strategies, munodeficiency virus type 1 strain. J Infec Dis
1998. Abstract 51, 35. 1996;174:962 – 8.
Peng C, Ho BK, Chang TW, Chang NT. Role of human Schmit J-C, Van Laethem K, Ruiz L, Hermans P, Sprecher S,
immunodeficiency virus type 1-specif protease in core Sonnerborg A, Leal M, Harrer T, Clotet B, Arendt V,
protein maturation and viral infectivity. J Virol Lissen E, Witvrouw M, Desmyter J, De Clercq E, Van-
1989;63:2550 – 6. damme A. Multiple dideoxynucleoside analogue-resistant
Perelson AS, Neumann AU, Markowitz M, Leonard JM, Ho (MddNR) HIV-1 strains isolated from patients from differ-
DD. HIV-1 dynamics in vivo: virion clearance rate, in- ent European countries. AIDS 1998;12:2007 – 15.
fected cell life-span, and viral generation time. Science Schuurman R, Demeter L, Reichelderfer P, Tijnagel J, de
1996;271:1582– 6. Groot T, Boucher C. Worldwide evaluation of DNA se-
Petropoulos CJ, Parkin NT, Limoni KL, Lie YS, Wrin T, quencing approaches for identification of drug resistance
Huang W, Tian H, Smith D, Winslow GA, Capon DJ, mutations in the human immunodeficiency virus type 1
Whitcomb JM. A novel phenotypic drug susceptibility reverse transcriptase. J Clin Microbiol 1999;37:2291 – 6.
assay for human immunodeficiency virus type 1. Antimi- Shafer RW, Kozal MJ, Winters MA, Iversen AKN, Katzen-
crob Agents Chemother 2000;44:920 –8. stein DA, Ragni MV, Meyer WA 3rd, Gupta P, Rasheed S,
Pierson T, McArthur J, Siliciano RF. Reservoirs for HIV-1: Coombs R, Katzman M, Fiscus S, Merigan TC. Combina-
mechanisms for viral persistence in the presence of antivi- tion therapy with zidovudine and didanosine selects for
ral immune responses and antiretroviral therapy. Annu drug-resistant human immunodeficiency virus type 1
Rev Immunol 2000;18:665 – 708. strains with unique patterns of pol gene mutations. J Infect
Puchhammer-Stockl E, Schmied B, Mandl CW, Vetter N, Dis 1994;169:722 –9.
Heinz FX. Comparison of line probe assay (LIPA) and Shafer RW, Iversen AKN, Winters MA, Aguiniga E, Katzen-
sequence analysis for detection of HIV-1 drug resistance. J stein DA, Merigan TC. AIDS Clinical Trials Group 143
Med Virol 1999;57:283 –9. Virology Team. Drug resistance and heterogeneous long-
Pyra H, Boni J, Schupbach J. Ultrasensitive retrovirus detec- term virologic responses of human immunodeficiency virus
tion by a reverse transcriptase assay based on product-en- type 1-infected subjects to zidovudine and didanosine com-
hancement. Proc Natl Acad Sci USA 1994;91:1544 –8. bination therapy. J Infect Dis 1995;172:70 – 8.
Qari SH, Respess R, Weinstock H, Beltrami E, Hertogs K, Shirasaka T, Yarchoan R, O’Brien C, Husson RN, Anderson
Larder BA, Petropoulos CJ, Hellmann N, Heneine W. A BD, Kojima E, Shimada T, Broder S, Mitsuya H. Changes
comparative analysis of Virco Antivirogram and Virologic in drug sensitivity of human immunodeficiency virus type 1
Phenosense phenotypic assays for drug susceptibility of during therapy with azidothymidine, dideoxycytidine, and
HIV-1. In: 3th International Workshop on HIV Drug dideoxyinosine: an in vitro comparative study. Proc Natl
Resistance and Treatment Strategies, 2000; abstract 62. Acad Sci USA 1993;90:562 – 6.
Richman DD, Havlir D, Corbeil J, Looney D, Ignacio C, Shirasaka T, Kavlick MF, Ueno T, Gao W, Kojima E,
Spector SA, Sullivan J, Cheesermran S, Barringer K, Alcaide ML, ChokekiJchai S, Roy BM, Arnold E,
212 J.G. García-Lerma, W. Heneine / Journal of Clinical Virology 21 (2001) 197–212

Yarchoan R, Mitsuya H. Emergence of human im- B, Van Ranst M, Desmyter J, De Clercq E, Vandarnme A.
munodeficiency virus type 1 variants with resistance to Patients HIV-1 strains carrying the multiple nucleoside
multiple dideoxynucleosides in patients receiving therapy resistance mutations are crossresistant to abacavir. AIDS
with dideoxynucleosides. Proc Natl Acad Sci USA 2000;14:469 – 71.
1995;92:2398 – 402. Vázquez-Rosales JG, Garcia-Lerma JG, Yamamoto S, Switzer
Silver J, Maudru T, Fujita Y, Repaske R. An RT-PCR assay WM, Havlir D, Folks TM, Richman DD, Heneine W.
for the enzyme activity of reverse transcriptase capable of Rapid screening of phenotypic resistance to nevirapine by
detecting single virions. Nucleic Acids Res 1993;21:3593 – 4. direct analysis of HIV type 1 reverse transcriptase activity
Spence RA, Kati WM, Anderson KS, Johnson KA. Mecha- in plasma. AIDS Res Hum Retrovir 1999;15:1191 – 200.
nisms of inhibition of HIV-1 reverse transcriptase by non- Winters MA, Coolley KL, Girard YA, Levee DJ, Hamdan H,
nucleoside inhibitors. Science 1995;267:988 –93. Shafer RW, Katzenstein DA, Merigan TC. A 6-basepair
St Clair MH, Martin JL, Tudor-Williams G, Bach MC, Vavro insert in the reverse transcriptase gene of human im-
CL, King DM, Kellam P, Kemp SD, Larder BA. Resis- munodeficiency virus type 1 confers resistance to multiple
tance to ddI and sensitivity to AZT induced by a mutation nucleoside inhibitors. J Clin Invest 1998;102:1769 – 75.
in HIV-1 reverse transcriptase. Science 1991;253:1557 –9. Witvrouw M, Pannecouque C, Van Laethem K, Desmyter J,
Stuyver L, Wyseur A, Rombout A, Louwagie J, Scarcez T, De Clercq E, Vandarnme AM. Activity of non-nucleoside
Verhotstede C, Rimland D, Schinazi RF, Rossau R. Line reverse transcriptase inhibitors against HIV-2 and SIV.
probe assay for rapid detection of drug-selected mutations
AIDS 1999;13:1477 – 83.
in human immunodeficiency virus type 1 reverse transcrip-
Wu JC, Warren TC, Adams J, Proudfoot J, Skiles J, Ragha-
tase gene. Antimicrob Agents Chemother 1997;41:284 –91.
van P, Perry C, Potocki I, Farina PF, Grob PM. A novel
Stuyver L, Wyseur A, Verpooten G, Rossau R. Line probe
dipyridodiazepinone inhibitor of HIV-1 reverse transcrip-
assay for detecting mutations in HIV-1 reverse transcrip-
tase acts through a non substrate binding site. Biochem-
tase. In: Kinchington D, Schinazi RF, editors. Methods in
istry 1991;30:2022 – 6.
Molecular Medicine: Antiviral Methods and Protocols,
Yamamoto S, Folks TM, Heneine W. Highly sensitive qualita-
vol. 24. New Jersey: Humana Press, 1999:259 – 82.
tive and quantitative detection of reverse transcriptase
Tisdale M, Kemp D, Parry NR, Larder BA. Rapid in vitro
activity: optimization, validation, and comparative analysis
selection of human immunodeficiency virus type 1 resistant
to 3%-thiacytidine inhibitors due to a mutation in the with other detection systems. J Virol Methods
YMDD region of the reverse transcriptase. Proc Natl Acad 1996;61:135 – 43.
Sci USA 1993;90:5653 –6. Yarchoan R, Mitsuya H, Thomas RV, Pluda JM, Hartman
Ueno T, Shirasaka T, Mitsuya H. Enzymatic characterization NF, Perno CF, Marczyk KS, Allain JP, Johns DG, Broder
of human immunodeficiency virus type 1 reverse transcrip- S. In vivo activity against HIV and favorable toxicity
tase resistant to multiple 2%, 3%-dideoxynucleoside 5%- profile of 2%,3%-dideoxyinosine. Science 1989;245:412 – 5.
triphosphates. J Biol Chem 1995;270:23605 –11. Young SD, Britcher SF, Tran LO, Payne LS, Lumma WC,
Vacca JP, Dorsey BD, Schleif WA, Levin RB, McDaniel SL, Lyle TA, Huff JR, Anderson PS, Olsen DB, Carroll SS.
Darke PL, Zugay J, Quintero JC, Blahy OM, Roth E, L-743, 726 (DN4P-266): a novel, highly potent nonnu-
Sardana VV, Schlabach AJ, Graham PI, Condra JH, Gtlib cleoside inhibitor of the human immunodeficiency virus
L, Holloway MK, Lin J, Chen I-W, Vastag K, Ostovich D, type 1 reverse transcriptase. Antimicrob Agents Cher-
Andersen PS, Emini EA, Huff JR. L-735,524: an orally nother 1995;39:2602 – 5.
bioavailable human immunodeficiency virus type 1 Zhang D, Caliendo AM, Eron JJ, DeVore KM, Kaplan JC,
protease inhibitor. Proc Natl Acad Sci USA 1994;91:4096 – Hirsch MS, D’Aquila RT. Resistance to 2%,3%-dideoxycy-
100. tidine conferred by a mutation in codon 65 of the human
Van Laethem K, Witvrouw M, Balzarini J, Schmit JC, immunodeficiency virus type 1 reverse transcriptase. An-
Sprecher S, Hermans P, Leal M, Harrer T, Ruiz L, Clotet timicrob Agents Chemother 1994;38:282 – 7.

You might also like